EP2655407A2 - Variants de ligands de la famille du tnf - Google Patents

Variants de ligands de la famille du tnf

Info

Publication number
EP2655407A2
EP2655407A2 EP11813430.3A EP11813430A EP2655407A2 EP 2655407 A2 EP2655407 A2 EP 2655407A2 EP 11813430 A EP11813430 A EP 11813430A EP 2655407 A2 EP2655407 A2 EP 2655407A2
Authority
EP
European Patent Office
Prior art keywords
variant
tnf family
family ligand
ligand
dimer
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP11813430.3A
Other languages
German (de)
English (en)
Inventor
Wilhelmus Johannes Quax
Vicente R TUR
Luis Serrano
Albert Martinus Van Der Sloot
Robbert H. COOL
Aart H. G. Van ASSEN
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Europaisches Laboratorium fuer Molekularbiologie EMBL
Institucio Catalana de Recerca i Estudis Avancats ICREA
Fundacio Privada Centre de Regulacio Genomica CRG
Rijksuniversiteit Groningen
Original Assignee
Europaisches Laboratorium fuer Molekularbiologie EMBL
Institucio Catalana de Recerca i Estudis Avancats ICREA
Fundacio Privada Centre de Regulacio Genomica CRG
Rijksuniversiteit Groningen
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Europaisches Laboratorium fuer Molekularbiologie EMBL, Institucio Catalana de Recerca i Estudis Avancats ICREA, Fundacio Privada Centre de Regulacio Genomica CRG, Rijksuniversiteit Groningen filed Critical Europaisches Laboratorium fuer Molekularbiologie EMBL
Priority to EP11813430.3A priority Critical patent/EP2655407A2/fr
Publication of EP2655407A2 publication Critical patent/EP2655407A2/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/525Tumour necrosis factor [TNF]

Definitions

  • the Tumor Necrosis Factor ligand (TNF) family is a family of ligands which are involved in a wide range of biological activities, including cell proliferation and apoptosis. There is a complex balance between immunostimulatory and immunoregulatory functions within this family that ensures that an individual is capable of appropriate immune responses. Genetic polymorphisms or other mutations in the TNF ligand receptor family can result in deregulation of immune homeostasis, which is implicated in pathogenesis. For this reason, the TNF family represents a prime target for therapeutic intervention.
  • TNF family of ligands interacts with its cognate receptor(s) to trigger a number of signalling pathways that are important for immune tolerance, in addition to providing both protective and pathogenic effects on tissues (35,36,37).
  • ligands such as Receptor Activator of NF-Kappa Beta-Ligand (RANKL), TNF-related apopto sis-inducing ligand (TRAIL), B-Cell Activating Factor (BAFF), A Proliferation-inducing Ligand (APRIL), TNFalpha, CD30L, CD40L, FasL, Light, and Tumor necrosis factor-like Weak inducer of apoptosis (Tweak), which are implicated in disease conditions such as rheumatoid arthritis, autoimmune diabetes, systemic lupus erythematosus (SLE), Sjorgen's syndrome, experimental autoimmune encephalomyelitis (EAE), inflammatory bowel disease (IBD), autoimmune
  • RIB
  • RANKL is particularly implicated in disorders associated with reduced bone density, such as osteoporosis, bone lesions due to rheumatoid arthritis (RA), Paget's disease and malignancy induced bone disease because signalling through RANKL results in increased production of bone-resorbing osteoclast cells. Binding of RANKL to its cognate receptor Receptor Activator of NF-Kappa Beta (RANK) expressed on osteoclast progenitor cells is crucial for the differentiation of these progenitor cells into mature osteoclasts, while binding of RANKL to RANK expressed on mature osteoclasts prevents these cells undergoing apoptosis and stimulates their adherence to bone cells (5). Increased signalling through RANKL and the production of an increased number of bone-resorbing osteoclasts disrupts the delicate homeostatic balance between osteoclasts and bone tissue producing osteoblasts, leading to disorders associated with reduced bone density, such as those mentioned above.
  • RANK NF-Kappa Beta
  • RANKL has been implicated in breast cancer. Here it is responsible for proliferative changes to the mammary epithelium which can lead to tumorigenesis (45,46). A soluble form of the RANKL extracellular domain is also produced by several types of tumour cells, including myeloma and metastatic breast cancer and by activated T lymphocytes in rheumatoid arthritis (5). RANKL has also been shown to promote the migration of RANK expressing tumor cells to bone tissue (48, 49, 50) and to control the development of progestin sensitive breast cancer (45, 46). The inventors therefore hypothesise that inhibiting the interaction of RANKL with its cognate receptor RANK could be used for the treatment of cancer and other diseases.
  • TNF ligand family members consist of anti-parallel ⁇ -sheets, which self-trimerise into the homotrimeric active form of the ligand. Sequence homology is highest between the residues found at the trimer interface. Once formed, a ligand trimer binds three cognate receptors, with each receptor either binding in the groove between two adjacent ligands or binding directly to one of the ligands. (33,34).
  • TNF family ligands As well as binding to their cognate receptors in order to promote signalling, TNF family ligands also bind to naturally occurring decoy receptors. These decoy receptors are either membrane bound or soluble pseudo-receptors, to which the TNF ligand can bind, without activating cell signalling. Whilst bound to the decoy receptor, the TNF ligand is sequestered away from the active receptor, and receptor-mediated signalling is inhibited.
  • Osteoprotegerin is a soluble decoy receptor for RANKL. It is produced by osteoblasts and binds RANKL, thus preventing RANKL binding to and activating RANK, and resulting in a reduction of osteoclast activity.
  • DcRl and DcR2 are decoy receptors for TRAIL
  • DcR3 is a decoy receptor for FasL. These receptors sequester TRAIL or FasL, respectively, away from the active signalling receptors, effectively acting as endogenous competitive inhibitors.
  • One approach is to use the extracellular ligand binding domain of the ligand's endogenous receptor and fuse it with the Fc domain of IgG. These chimeric soluble receptors function as artificial decoy receptors and prevent the endogenous ligand binding to the endogenous receptor by sequestering it (51).
  • Etanercept Enbrel®
  • a similar approach has also been used with the RANKL decoy receptor OPG (44).
  • An alternative approach requires the administration of a trimeric TNF ligand mutant incapable of receptor binding but capable of subunit exchange with the wild-type TNF ligand. This may result in the formation of mixed mutant/endogenous ligand trimers which are either incapable of receptor binding; as are the trimeric mutant forms, or, if bound, incapable of activating the receptor once bound(47).
  • the trimeric wild-type ligand is essentially "poisoned" by the mutant variant and as a result, inactivated.
  • a drawback of this approach is the heterogeneity of the preparation containing different ratios of wild-type to mutant monomers in the trimer, which makes the effects unpredictable.
  • Another strategy which has been successfully applied is the administration of anti-ligand antibodies (e.g Denosumab, also known as AMG 162 and Prolia®/Xgeva®) (42,43). These antibodies bind to RANKL and prevent it from binding to its cognate receptor, therefore blocking receptor signaling.
  • anti-ligand antibodies e.g Denosumab, also known as AMG 162 and Prolia®/Xgeva®
  • blocking peptides have also been used to block signaling through the RANK/RANKL pathway by binding directly to RANK and blocking the ligand binding site (31,32).
  • the present invention relates to variants of TNF family ligands which have been mutated at the ligand trimerisation interface so that they are not capable of assembling into trimers, and either assemble into dimers or remain as monomers.
  • Such ligands bind to the TNF receptor but are unable to activate it, effectively functioning as competitive inhibitors.
  • the invention also relates to nucleic acids encoding the variants of TNF family ligands, vectors and host cells comprising the nucleic acid and methods for the treatment of diseases associated with aberrant signalling through a TNF receptor.
  • TNF ligand variants which are not capable of trimerisation have the advantage that they are structurally similar to wild-type ligands, and are therefore less prone to proteolytic degradation than blocking peptides.
  • the TNF ligand variants of the present invention therefore have potentially greater stability than blocking peptides due to a decreased rate of degradation and clearance from the body. Further, such a system exploits the TNF ligand signaling pathway in a manner distinct from pre-existing therapies as this approach targets the receptor and not the ligand and it can therefore be used alone or in combination with pre-existing therapies.
  • the present invention includes a variant of a TNF family ligand which is mutated such that it is not capable of assembling into a trimer, wherein the variant retains the ability to bind to one or more of its cognate receptor(s), but wherein binding to the receptor does not activate the receptor.
  • mutated encompasses substitution to any natural or non-natural amino acid residue, deletion, insertion and addition of amino acid residues.
  • the TNF ligand variant may include one or more post- translational modifications. Suitable modifications include pegylation, acetylation, formylation, alkylation such as methylation, and glycosylation, as well as labeling with fluorophores, radioisotopes, PET, etc. Such modifications may decrease immunogenicity, improve pharmacokinetics, and allow for certain specific applications of the variants (e.g. diagnostics). As discussed above, TNF ligand trimerisation is required to effect receptor trimerisation, which is in turn required for signalling.
  • a variant of a TNF family ligand which is incapable of trimerisation, but able to assemble into a dimer or remain monomeric, and still able to bind its TNF receptor, will effectively block its receptor and therefore inhibit downstream signalling.
  • the variant is therefore functioning as a competitive inhibitor.
  • the variant of a TNF family ligand may be capable of assembling into a dimer with another variant of the same TNF family ligand.
  • TNF ligand dimer (homodimer or heterodimer), which is capable of binding to its cognate receptor, but will not be capable of activating the receptor because receptor trimerisation cannot occur when there is only a ligand dimer present.
  • the ligand variant has at least 10 fold, at least 100 fold, at least 1000 fold or more higher affinity for assembling into a dimer than assembling into a trimer.
  • the ligand variant may have an affinity of at least 10 8 M, at least 10 9 M, at least 10 10 M or greater for its cognate receptor(s).
  • Affinity may be defined using the association constant (Ka), which is determined using the following formulae:
  • [C] is the concentration of the complex
  • [R] is the concentration of unbound receptor
  • [L] is the concentration of unbound ligand
  • the ligand variant dissociation constant (Ka) of at least 10 "8 M, at least 10 "9 M, at least 10 "10 M or less. Kd is determined using the following formula:
  • [C] is the concentration of the complex
  • [R] is the concentration of unbound receptor
  • [L] is the concentration of unbound ligand.
  • the present invention potentially ensures a faster response with a lesser dose, as it allows improvement of the Kd to provide stronger antagonists.
  • TNF family receptors bind to their cognate ligands in the groove formed between two ligands of the trimer (e.g. RANK, TNFR1 , TNFR2, FAS, CD40, CD27, CD30, DR4 and DR5).
  • trimer e.g. RANK, TNFR1 , TNFR2, FAS, CD40, CD27, CD30, DR4 and DR5
  • a dimer will be required in order to stably bind, and therefore block, a single TNF receptor. Only one receptor will be bound by such a dimer due to the presence of only one cleft in which a receptor can be bound. The receptors will therefore be blocked, and no signalling will occur.
  • TNF family receptors bind their cognate ligands within the structure of the TNF-ligand monomer, generally the solvent exposed surface of the TNF ligand monomer (e.g. APRIL and BAFF).
  • the dimer is formed of two variant TNF ligands that bind to two TNF receptors.
  • receptor activation will not occur due to the absence of the third ligand, which is required for receptor trimerisation.
  • the TNF family ligand from which the variant is derived may be selected from the group consisting of RANKL (human accession no. AAB86811, mouse accession no. 035235), TRAIL (human accession no. P50591), APRIL (human accession no. BAE16556), BAFF (human accession no: Q9Y275), TNFalpha (human accession no. NP 000585), TNFbeta (human accession no. P01374), CD30L (human accession no. NP 001235; also NM 001244), CD40L (human accession no. NP 000065), FasL (human accession no. NP 000630), Light (human accession no.
  • RANKL human accession no. AAB86811, mouse accession no. 035235
  • TRAIL human accession no. P50591
  • APRIL human accession no. BAE16556
  • BAFF human accession no: Q9Y275
  • TNFalpha human accession no
  • the TNF family ligand may be of mammalian origin. More specifically, the TNF family ligand may be human, camel, dog, cat, horse, cow, pig, sheep, camelid, mouse, rat, rabbit, hamster, guinea pig, pig, sheep, and so on.
  • the variant of a TNF family ligand may be mutated at one or more positions. In certain embodiments, the variant may be mutated at 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more positions. It is preferred that the ligand variant is a soluble ligand variant. In this embodiment, the ligand may have been mutated to delete the transmembrane domain.
  • the ligand variant may comprise or consist of amino acid residues 159-317 or 161-317, and variants around this sequence. A RANKL ligand variant comprising or consisting of amino acids 161-317 is preferred. It will be clear to the skilled reader how to design equivalent soluble ligand variants from other TNF ligand family members and specific examples are given in more detail below.
  • the inventors have illustrated the principle of preparing TNF ligand variants which are incapable of forming trimers using RANKL.
  • the TNF family ligand is RANKL.
  • TNF family ligands including TRAIL, APRIL, BAFF, TNFalpha, TNFbeta, CD30L, CD40L, FasL, Light, and Tweak, GITRL, EDA-A1, EDA-A2 and OX40L.
  • the RANKL variant may comprise or consist of a mutation at one or more of positions 169, 195, 213, 230, 257, 272, and 280 in the human RANKL sequence, for example, at 2 or 3 or 4 or 5 or 6 or all 7 of these positions (representative example combinations in human RANKL include 195 and 272; 213, 257 and 280; 213 and 280; 169, 230 and 272; 169, 213, 230, 257 and 280).
  • TNF ligand variants mutated at equivalent positions in orthologous proteins.
  • these positions are 168, 194, 212, 229, 256, 271, and 279.
  • Mutations at such positions may be to any natural or non-natural amino acid, although mutations to more hydrophilic amino acids are preferred because residues positioned at the trimer interface will not be surface exposed following formation of a trimer, but at least some will be surface exposed for a TNF ligand variant incapable of forming a trimer.
  • a human RANKL variant may comprise or consists of one or more of the mutations T169V, K195D, F213Y, D230K, K257D, F272Y and F280Y.
  • these mutations are T168V, K194D, F212Y, D229K, K256D, F271Y and F279Y. Therefore, the variant may comprise or consist of 1, 2, 3, 4, 5, 6 or 7 of these mutations.
  • the variant may comprise a mutation at position 169 of the human RANKL sequence or at a position equivalent thereto in orthologous proteins. In the murine sequence, this position is 168.
  • the variant may comprise the mutation T169V in the human RANKL sequence or at a position equivalent thereto in orthologous proteins.
  • this mutation is T168V.
  • the variant may comprise a mutation at position 195 of the human RANKL sequence or at a position equivalent thereto in orthologous proteins. In the murine sequence, this position is 194.
  • the variant may comprise the mutation K195D in the human RANKL sequence.
  • this mutation is K194D.
  • the variant may comprise a mutation at position 213 of the human RANKL sequence or at a position equivalent thereto in orthologous proteins. In the murine sequence, this position is 212.
  • the variant may comprise the mutation F213 Y in the human RANKL sequence or at a position equivalent thereto in orthologous proteins.
  • this mutation is F212Y.
  • the variant may comprise a mutation at position 230 of the human RANKL sequence or at a position equivalent thereto in orthologous proteins. In the murine sequence, this position is 229.
  • the variant may comprise the mutation D230K in the human RANKL sequence or at a position equivalent thereto in orthologous proteins.
  • this mutation is S229K.
  • the variant may comprise a mutation at position 257 of the human RANKL sequence or at a position equivalent thereto in orthologous proteins. In the murine sequence, this position is 256.
  • the variant may comprise the mutation K257D in the human RANKL sequence or at a position equivalent thereto in orthologous proteins. In the murine sequence this mutation is K256D. In one embodiment the variant may comprise a mutation at position 272 of the human RANKL sequence or at a position equivalent thereto in orthologous proteins. In the murine sequence, this position is 271.
  • the variant may comprise the mutation F272Y in the human RANKL sequence or at a position equivalent thereto in orthologous proteins.
  • this mutation is F271Y.
  • the variant may comprise a mutation at position 280 of the human RANKL sequence or at a position equivalent thereto in orthologous proteins. In the murine sequence, this position is 279.
  • the variant may comprise the mutation F280Y in the human RANKL sequence or at a position equivalent thereto in orthologous proteins.
  • this mutation is F279Y.
  • the variant may also comprise one or more additional mutations which improve the solubility or stability of the dimer. Therefore, in one embodiment, the variant may comprise a mutation at one or more of positions 207, 221 or 247 in the human RANKL sequence, or at a position equivalent thereto in orthologous proteins. In the murine sequence these position are 206, 220 and 246. Mutations at these positions may be to any natural or non-natural amino acid, although mutation to a more hydrophilic amino acids is preferred to improve stability. In particular, the mutations I207R, C221 S and I247E in the human RANKL sequence (R206R, C220S and I246E in the murine RANKL sequence) are preferred. The variant may comprise 1, 2 or 3 of these mutations.
  • the RANKL variant may comprise or consist of mutations at positions K195, C221 and F272 in the human RANKL sequence, or equivalent positions in orthologous proteins (in the mouse these are K194, C220 and F271).
  • the RANKL variant may comprise or consist of the mutations K195D, C221S and F272Y in the human RANKL sequence, or equivalent mutations at equivalent positions in orthologous proteins (in the mouse these are K194D, C220S and F271Y).
  • This variant is herein referred to as hDl-lA and forms a monomer in the hDl-1 dimer, with monomer hDl-lB (the equivalent in the mouse is referred herein as monomer mD 1 A, and together with monomer mD 1 B , forms dimer mD 1 ) .
  • the RANKL variant may comprise or consist of mutations at positions F213, K257 and F280 in the human RANKL sequence, or equivalent positions in orthologous proteins (in the mouse these are F212, K256 and F279).
  • the RANKL variant may comprise or consist of the mutations F213Y, K257D and F280Y in the human RANKL sequence, or equivalent mutations at equivalent positions in orthologous proteins (in the mouse these are F212Y, K256D and F279Y).
  • This variant is herein referred to as hDl-lB and forms one monomer in the hDl-1 dimer, with monomer hDl-lA (the equivalent in the mouse is referred herein as monomer mDlB, and together with monomer mDIA, forms dimer mDl).
  • the RANKL variant may comprise or consist of mutations at positions K195, 1207, C221 and F272 in the human RANKL sequence, or equivalent positions in orthologous proteins (in the mouse these are K194, R206, C220 and F271).
  • the RANKL variant may comprise or consist of the mutations K195D, I207R, C221S and F272Y in the human RANKL sequence, or equivalent mutations at equivalent positions in orthologous proteins (in the mouse these are K194D, R206R, C220S and F271Y).
  • This variant is herein referred to as hDl-2A and forms one monomer in the hDl-2 dimer, with monomer hDl-2B.
  • the RANKL variant may comprise or consist of mutations at positions 1207, F213, K257 and F280 in the human RANKL sequence, or equivalent positions in orthologous proteins (in the mouse these are R206, F212, K256 and F279).
  • the RANKL variant may comprise or consist of the mutations I207R, F213Y, K257D and F280Y in the human RANKL sequence, or equivalent mutations at equivalent positions in orthologous proteins (in the mouse these are R206R, F212Y, K256D and F279Y).
  • This variant is herein referred to as hDl-2B and forms one monomer in the hD 1 -2 dimer, with monomer hD 1 -2 A.
  • the RANKL variant may comprise or consist of mutations at positions K195, C221, 1247 and F272 in the human RANKL sequence, or equivalent positions in orthologous proteins (in the mouse these are K194, C220, 1246 and F271).
  • the RANKL variant may comprise or consist of the mutations K195D, C221S, I247E and F272Y in the human RANKL sequence, or equivalent mutations at equivalent positions in orthologous proteins (in the mouse these are K194D, C220S, I246E and F271Y).
  • This variant is herein referred to as hDl-3A and forms one monomer in the hDl-3 dimer, with monomer hDl-3B.
  • the RANKL variant may comprise or consist of mutations at positions F213, 1247, K257 and F280 in the human RANKL sequence, or equivalent positions in orthologous proteins (in the mouse these are F212, 1246, K256 and F279).
  • the RANKL variant may comprise or consist of the mutations F213Y, I247E, K257D and F280Y in the human RANKL sequence, or equivalent mutations at equivalent positions in orthologous proteins (in the mouse these are F212Y, I246E, K256D and F279Y).
  • This variant is herein referred to as hDl-3B and forms one monomer in the hDl-3 dimer, with monomer hDl-3A.
  • the RANKL variant may comprise or consist of mutations at positions T169, C221, D230 and F272 in the human RANKL sequence, or equivalent positions in orthologous proteins (in the mouse these are T168, C220, S229 and F271).
  • the RANKL variant may comprise or consist of the mutations T169V, C221S, D230K and F272Y in the human RANKL sequence, or equivalent mutations at equivalent positions in orthologous proteins (in the mouse these are T168V, C220S, S229K and F271Y).
  • hD2-lA This variant is known herein as hD2-lA and forms the hD2- 1 dimer, together with monomer hD2-lB (the equivalent in the mouse is referred herein as monomer mD2A, and together with monomer mD2B, forms dimer mD2).
  • the RANKL variant may comprise or consist of mutations at positions T169, F213, D230, K257 and F280 in the human RANKL sequence or equivalent positions in orthologous proteins (in the mouse these are T168, F212, S229, K256 and F279).
  • the RANKL variant may comprise or consist of the mutations T169V, F213Y, D230K, K257D and F280Y in the human RANKL sequence or equivalent mutations at equivalent positions in orthologous proteins (in the mouse these are T168V, F212Y, S229K, K256D and F279Y).
  • This monomer is known herein as hD2-lB, and forms the dimer hD2-l, together with monomer h2-lA (the equivalent in the mouse is referred herein as mD2B, and forms the dimer mD2, together with monomer mD2A).
  • the RANKL variant may comprise or consist of mutations at positions T169, 1207, C221, D230 and F272 of the human RANKL sequence, or equivalent positions in orthologous proteins (in the mouse these are T168, R206, C220, S229 and F271).
  • the RANKL variant may comprise or consist of the mutations T169V, I207R, C221S, D230K and F272Y in the human RANKL sequence, or equivalent mutations at equivalent positions in orthologous proteins (in the mouse these are T168V, R206R, C220S, S229K and F271Y).
  • This monomer is known herein as hD2- 2A, and forms the dimer hD2-2, together with monomer hD2-2B.
  • the RANKL variant may comprise or consist of mutations at positions T169, 1207, F213, D230, K257 and F280 in the human RANKL sequence or equivalent positions in orthologous proteins (in the mouse these are T168, R206, F212, S229, K256 and F279).
  • the RANKL variant may comprise or consist of the mutations T169V, I207R, F213Y, D230K, K257D and F280Y in the human RANKL sequence or equivalent mutations at equivalent positions in orthologous proteins (in the mouse these are T168V, R206, F212Y, S229K, K256D and F279Y).
  • This monomer is known herein as hD2-2B, and forms the dimer hD2-2, together with monomer hD2-2A.
  • the RANKL variant may comprise or consist of mutations at positions T169, C221, D230, 1247 and F272 in the human RANKL sequence, or equivalent positions in orthologous proteins (in the mouse these are T168, C220, S229, 1246 and F271).
  • the RANKL variant may comprise or consist of the mutations T169V, C221S, D230K, I247E and F272Y in the human RANKL sequence, or equivalent mutations at equivalent positions in orthologous proteins (in the mouse these are T168V, C220S, S229K, I246E and F271Y).
  • This monomer is known herein as hD2- 3A, and forms the dimer hD2-3, together with monomer hD2-3B.
  • the RANKL variant may comprise or consist of mutations at positions T169, F213, D230, 1247, K257 and F280 of the human RANKL sequence or equivalent positions in orthologous proteins (in the mouse these are T168, F212, S229, 1246, K256 and F279).
  • the RANKL variant may comprise or consist of the mutations T169V, F213Y, D230K, I247E, K257D and F280Y in the human RANKL sequence or equivalent mutations at equivalent positions in orthologous proteins (in the mouse these are T168V, F212Y, S229K, I246E, K256D and F279Y).
  • This monomer is known herein as hD2-3B, and forms the dimer hD2-3, together with monomer hD2-3A.
  • the RANKL variant may comprise or consist of mutations at positions C221 and F272 of the human RANKL sequence or equivalent positions in orthologous proteins (in the mouse these are C220 and F271).
  • the RANKL variant may comprise or consist of the mutations C221S and F272Y in the human RANKL sequence or equivalent mutations at equivalent positions in orthologous proteins (in the mouse these are C220S and F271Y).
  • the mouse monomer is known herein as mDl 'A, and forms the dimer mDl ', together with monomer mDl 'B.
  • the RANKL variant may comprise or consist of mutations at positions F213, K257 and F280 of the human RANKL sequence or equivalent positions in orthologous proteins (in the mouse these are F212, K256 and F279).
  • the RANKL variant may comprise or consist of the mutations F213Y, K257D and F280Y in the human RANKL sequence or equivalent mutations at equivalent positions in orthologous proteins (in the mouse these are F212Y, K256D and F279Y).
  • the mouse monomer is known herein as mDl 'B, and forms the dimer mDl ', together with monomer mDl ⁇ .
  • the present invention includes a variant of a TNF ligand variant having mutations at positions equivalent to those discussed above in relation to RANKL. Such equivalent positions are detailed in Table 1, below. These positions are derived from figures 31 and 32.
  • Human RANKL (hRANKL) has very high sequence similarity to mRANKL (>88% sequence identity, Figure 22), and all but one amino acids at the positions recited above are conserved at least between mRANKL and hRANKL. Human counterparts to the mRANKL variants described herein are therefore highly similar in sequence to the mRANKL variants, and are included within the scope of the invention. Mutations Equivalent mutations positions in other TNF family ligands
  • the invention includes the TNF ligand variants of SEQ ID NOs: 33-
  • the invention embraces any TNF superfamily member (including Human TRAIL, Human BAFF, human TNFa, human TNFp, Human CD40L, Human APRIL, Human OX40L or Human GITRL) with a mutation at a position set out in the above table. Mutations at multiple positions are also aspects of the invention, so TNF ligand superfamily members may have 1 , 2, 3, 4, 5, 6, 7, 8 or more mutations at the 10 positions set out in the table below.
  • TNF superfamily member including Human TRAIL, Human BAFF, human TNFa, human TNFp, Human CD40L, Human APRIL, Human OX40L or Human GITRL
  • the TNF family ligand variants may be synthesised with a tag sequence to aid purification.
  • tag sequences can be fused to either one or both of the monomers comprising the heterodimer.
  • the tag may be linked to either the N terminus or the C terminus of the 15 protein, and may be linked either covalently or non-covalently. Preferably, the tag is linked covalently.
  • This tag may be one of any number of suitable tags, as will be appreciated by one of skill in the art, such as a histidine tag, a FLAG tag, a biotin or streptavidin tag; preferably, the tag is a histidine tag, such as one with the sequence MGSSHHHHHHSQDP (SEQ ID NO: 124).
  • the component monomers of the dimer may both be tagged, or just one may be tagged. If both are tagged, different tags may be used.
  • the invention includes a dimer comprising or consisting of two of the variants of TNF family ligands of the invention.
  • a dimer may include any two TNF ligand variants of the same ligand described above.
  • the dimer comprises or consists of two of the RANKL variants discussed above.
  • the dimer of two variants of TNF family ligands may be a heterodimer.
  • the two variants included in the dimer may therefore contain different mutations to each other, relative to the wild-type sequence.
  • Representative examples are the hDl-1, hDl-2, hDl-3, hD2-l, hD2-2 and hD2-3 dimers described above, which are preferred examples of dimers according to this aspect of the invention.
  • Preferred dimer forms in the murine sequence are the mDl, mDl ' and mD2 dimers.
  • the two ligand variants may be present as a single peptide chain.
  • sequences of the two ligand variants may be directly linked, or they may be linked through a linker sequence comprising or consisting of, for example, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50 or more amino acids.
  • the dimer may comprise or consist of a variant comprising or consisting of mutations at positions C221 and F272 of the human RANKL sequence or equivalent positions (mouse is C220 and F271), and a variant comprising or consisting of mutations at positions F213, K257 and F280 of the human RANKL sequence or equivalent positions (mouse is F212, K256 and F279).
  • the dimer may comprise or consist of a variant comprising or consisting of the mutations C221S and F272Y of the human RANKL sequence or equivalent mutations at equivalent positions (mouse is C220S and F271Y), and a variant comprising or consisting of the mutations F213Y, K257D and F280Y of the human RANKL sequence or equivalent mutations at equivalent positions (mouse is F212Y, K256D and F279Y). In the mouse, this is termed mDl ' herein.
  • the dimer may comprise or consist of a variant comprising or consisting of mutations at positions T169, C221, D230 and F272 of the human RA KL sequence or equivalent positions (mouse is T168, C220, S229 and F271) and a variant comprising or consisting of mutations at positions T169, F213, D230, K257 and F280 of the human RANKL sequence or equivalent positions (mouse is T168, F212, S229, K256 and F279).
  • the dimer may comprise or consist of a variant comprising or consisting of the mutations T169V, C221S, D230K and F272Y of the human RANKL sequence or equivalent mutations (mouse is T168V, C220A, S229K and F271 Y) at equivalent positions and a variant comprising or consisting of the mutations T169V, F213Y, D230K, K257D and F280Y of the human RANKL sequence or equivalent mutations at equivalent positions (in mouse these are T168V, F212Y, S229K, K256D and F279Y ).
  • This dimer is herein termed hD2-l in the human and mD2 in the mouse.
  • the TNF ligand variant dimer may comprise or consist of two of the variants of SEQ ID NOs: 35-110, with the proviso that the two variant monomers forming the dimer are variants of the same TNF ligand.
  • the two ligand variants are SEQ ID NOs: 35 and 36, SEQ ID NOs: 79 & 80, SEQ ID NOs: 81 & 82, SEQ ID NOs: 37 and 38, SEQ ID NOs: 83 & 84, SEQ ID NOs: 85 & 86, SEQ ID NOs: 87 & 88 SEQ ID NOs: 39 and 40, SEQ ID NOs: 89 & 90, SEQ ID NOs: 39 & 90, SEQ ID NOs: 89 & 40, SEQ ID NOs: 91 & 93, SEQ ID NOs: 92 & 94, SEQ ID NOs: 91 & 94, SEQ ID NOs: 92 & 93, SEQ ID NOs: 95 & 97, SEQ ID NOs: 96 & 98, SEQ ID NOs: 95 & 98, SEQ ID NOs: 96 & 97 SEQ ID NOs: 41 and 42, SEQ ID NOs: 85
  • the variant of a TNF family ligand may not be capable of assembling into a dimer with other variants of the same TNF ligand.
  • the variant will not be able to bind a receptor which binds within the cleft formed between two ligands because a ligand dimer will not form and there will therefore be no cleft into which a receptor can bind.
  • the ligand variant binds its cognate receptor on the solvent exposed surface rather than in the cleft between two adjacent ligand monomers, as shown in figure 30. This will enable the ligand variant to bind to its cognate receptor without any requirement for the ligand variant to dimerise.
  • TNF family ligands which bind in such a mode to their cognate receptor, and are therefore useful within this embodiment of the invention are APRIL, BAFF and most likely Tweak.
  • TNF ligand variants of the invention bind to the TNF receptor but are unable to activate it.
  • the TNF ligand variants of the invention are considered to "bind to the TNF receptor" if they demonstrate more than 50% of the receptor binding observed with the wild-type ligand at saturating concentration.
  • the ligand variants may demonstrate more than 60%, more than 70%, more than 80%, more than 90%, more than 91%, more than 92%, more than 93%, more than 94%, more than 95%, more than 96%, more than 97%, more than 98%, more than 99%, more than 99.5%, more than 99.6%, more than 99.7%, more than 99.8%, more than 99.9%, more than 99.95%, or more than 99.99% of the receptor binding observed with the wild-type ligand.
  • the TNF ligand variants of the invention are considered "unable to activate the receptor" if they demonstrate less than 50% of the receptor activation observed with the wild-type ligand at saturating concentration.
  • the ligand variants may demonstrate less than 40%>, less than 30%>, less than 20%>, less than 10%>, less than 9%, less than 8%, less than 7%, less than 6%, less than 5%, less than 4%, less than 3%, less than 2%, less than 1%, less than 0.5%, less than 0.4%, less than 0.3%, less than 0.2%, less than 0.1%, less than 0.05%, or less than 0.01% of the receptor activation observed with the wild-type ligand.
  • receptor activation may be assessed by measuring the levels of signalling through the receptor.
  • the variant of a TNF family ligand according to the invention may have an increased binding affinity for one or more of its cognate receptor(s), compared to the wild-type TNF family ligand.
  • the ligand variant may have increased binding affinity for 1, 2, 3, 4, 5 or more of its cognate receptors.
  • the binding affinity for one or more of the cognate receptors may be increased by substituting one or more amino acids residues of the TNF family ligand to any other natural or non-natural amino acid, deleting one or more amino acid residues from the TNF family ligand or inserting one or more amino acid residues into the TNF family ligand.
  • binding affinity may be increased by substituting one or more of the amino acid residues present at the receptor binding interface of the ligand to any other natural or non-natural amino acid, or deleting any of the residues present at the receptor binding interface or inserting one or more additional residues at the receptor binding interface.
  • the ligand variant may have an increased binding affinity for one or more of its cognate target receptors, compared to the wild-type TNF family ligand.
  • a "target" receptor refers to a receptor through which signalling can take place, i.e. not a decoy receptor, and which should be inhibited in order to produce a cellular response.
  • Non-target receptors refer to active receptors which are not the main target of inhibition and/or decoy receptors.
  • affinity for the cognate target receptor may be increased by any of the methods discussed above.
  • the ligand variant may include one or more amino acid residue substitutions or deletions which decrease the affinity of the ligand variant for one or more if its cognate decoy receptors.
  • the ligand variant may include one or more amino acid residue substitutions, insertions or deletions which decrease the affinity of the ligand variant for one or more of its non-target cognate receptors.
  • a receptor selective effect may be useful in cases where it is beneficial to inhibit the signalling mediated by one cognate receptor but not the signalling mediated by the other receptor(s) of the ligand.
  • it may be beneficial to block TNFalpha mediated signalling via TNFR1 but leave TNFalpha mediated TNFR2 signalling unperturbed.
  • Such receptor selectivity is not possible, or at least not as readily possible, with the approaches used in the prior art.
  • the TNF ligand variants of the invention may have at least 50% sequence identity to any one of the TNF ligands variants described herein and to SEQ ID NOs: 35-110.
  • the variant may have at least 60%, at least 70%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, at least 99.5%, at least 99.9% or more sequence identity to SEQ ID NOs: 35-110.
  • the variant may retain the ability to bind its cognate receptor(s).
  • the invention also encompasses fragments of the TNF ligand variants described herein and of SEQ ID NOs: 35-110.
  • a fragment may be 7, 8, 9, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250, 300 or more amino acids in length.
  • the variant fragment may retain the ability to bind its cognate receptor(s).
  • the invention also encompasses TNF ligand variants as described herein which have a truncation at the N-terminus and/or the C-terminus.
  • the truncation may be 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200 or more amino acids in length.
  • the truncation may be an internal deletion with the same characteristics.
  • the variant truncate may retain the ability to bind its cognate receptor(s).
  • the ligand variant may be a soluble ligand variant. Therefore, the ligand may have been mutated to delete the transmembrane domain.
  • the ligand variant may therefore comprise or consist of amino acid residues 158-316 or 160-316 of murine RANKL, and variants around this sequence, such as those described above.
  • the ligand variant may comprise or consist of amino acid residues 159-317 or 161-317 of human RANKL (161-317 is preferred), and variants around this sequence, such as those described above.
  • the ligand variant may therefore comprise or consist of amino acid residues 114-281 of human TRAIL, and variants around this sequence, such as those described above.
  • the ligand variant may therefore comprise or consist of amino acid residues 114-250 of human APRIL, and variants around this sequence, such as those described above.
  • the ligand variant may therefore comprise or consist of amino acid residues 134-285 of human BAFF, and variants around this sequence, such as those described above.
  • the ligand variant may therefore comprise or consist of amino acid residues 84-233 of human TNF alpha, and variants around this sequence, such as those described above.
  • the ligand variant may therefore comprise or consist of amino acid residues 62-205 of human TNF beta, and variants around this sequence, such as those described above.
  • the ligand variant may therefore comprise or consist of amino acid residues 95-234 of human CD30L, and variants around this sequence, such as those described above.
  • the ligand variant may therefore comprise or consist of amino acid residues 114-261 of human CD40L, and variants around this sequence, such as those described above.
  • the ligand variant may therefore comprise or consist of amino acid residues 130-281 of human FASL, and variants around this sequence, such as those described above.
  • the ligand variant may therefore comprise or consist of amino acid residues 83-240 of human LIGHT, and variants around this sequence, such as those described above.
  • the ligand variant may therefore comprise or consist of amino acid residues 94-249 of human TWEAK, and variants around this sequence, such as those described above.
  • the ligand variant may therefore comprise or consist of amino acid residues 72-199 of human GITRL, and variants around this sequence, such as those described above.
  • the ligand variant may therefore comprise or consist of amino acid residues 230-391 of human EDA-A1, and variants around this sequence, such as those described above.
  • the ligand variant may therefore comprise or consist of amino acid residues 51-183 of human OX40L, and variants around this sequence, such as those described above.
  • a table showing preferred sequences, extracellular forms, and preferred fragments, is set out below for TNF ligands according to the invention.
  • variants around these preferred fragments are included as aspects of the invention, which may be extended or truncated at either or both ends, for example, by 1, 2, 5, 10 or more amino acids.
  • the ligand variants of the invention may be fused to a heterologous peptide. This may allow purification of the ligand variant during production, or may confer an additional therapeutic property to the variant for therapeutic or diagnostic use such as increasing retention time in the circulatory system by allow weak binding to abundant blood proteins such as serum albumin.
  • a particularly suitable heterologous peptide for purification is a hexahistidine tag or a Flag tag.
  • heterologous peptide For heterodimers comprising two differing ligand variants, the heterologous peptide maybe be fused to one of the monomers constituting the heterodimer or to both, alternatively two different heterologous peptide sequences may be fused to the two monomers constituting the heterodimer.
  • the invention includes a nucleotide sequence encoding a variant of a TNF family ligand according to the invention.
  • the nucleic acid may encode any of the variants, truncates or fragments described above.
  • the nucleic acid may be DNA including chromosomal DNA and cDNA or RNA, including mRNA.
  • the mRNA or DNA sequences may or may not include one or more introns.
  • the nucleic acid may comprise or consist of any one of SEQ ID NOs: 111-123. Also included within the scope of the invention are fragments of these nucleic acid sequences. Such a fragment may be 7, 8, 9, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250, 300, 400, 500 or more nucleotides in length.
  • the nucleic acid may include one or more regulatory sequences such as a promoter, an enhancer, an internal ribosomal entry site (IRES), a Kozak sequence or a ribosomal binding site (RBS) sequence.
  • regulatory sequences such as a promoter, an enhancer, an internal ribosomal entry site (IRES), a Kozak sequence or a ribosomal binding site (RBS) sequence.
  • the monomers comprising the heterodimeric variants might be encoded in two separate nucleic acid molecules.
  • One or more ligand variants of the same TNF ligand may be encoded by a single nucleic acid molecule.
  • the nucleic acid molecule may encode two ligand variants of the same ligand.
  • expression of the two ligand variants may be under the control of the same or different promoters, IRES sites or other regulatory sequences.
  • the invention includes a vector comprising a nucleotide sequence encoding any of the variants, fragments or truncates of the invention.
  • Vectors suitable for use in the method of the invention include plasmids and viruses (including both bacteriophage and eukaryotic viruses), as well as other linear or circular DNA carriers, such as those employing transposable elements or homologous recombination technology.
  • Particularly suitable viral vectors include baculovirus-, lentivirus-, adenovirus- and vaccinia virus- based vectors.
  • the invention includes a host cell comprising a nucleic acid or a vector encoding one or more of the ligand variants of the invention.
  • host cells include prokaryotic host cells such as bacterial cells, yeast host cells and mammalian host cells. Particularly preferred host cells include E.coli, CHO, HEK293 PerC7 cells, B. subtilis, insect cells, and yeast such as S. cerevisiae.
  • the invention encompasses complexes which comprise or consist of one or more TNF ligand variants and one or more cognate TNF family receptors.
  • the complex may comprise or consist of a dimer formed of two TNF ligand variants which are incapable of assembling into a trimer and one or two cognate TNF family receptors.
  • the complex may comprise or consist of one or two TNF ligand variants which are incapable of assembling into dimers or trimers and one or two cognate TNF family receptors.
  • the invention includes a transgenic animal which has been engineered to express one or more of the TNF family variants or dimers disclosed herein.
  • the animal may be any animal which can be engineered to express one or more TNF ligand variants, for example a mammal.
  • suitable animals include monkeys, mice, cows, sheep, rabbit, rat, hamster, guinea pig, goat, horses, donkey, pigs, cats, dogs and camels.
  • the invention includes a pharmaceutical composition comprising one or more of the variants of a TNF family ligand, the dimers, the nucleotide sequences, the vectors or the host cells of the invention.
  • the pharmaceutical composition may additionally comprise a pharmaceutically-acceptable carrier, excipient, diluent or buffer.
  • Suitable pharmaceutically acceptable carriers, excipients, diluents or buffers may include liquids such as water, saline, glycerol, ethanol or auxiliary substances such as wetting or emulsifying agents, pH buffering substances and the like. Excipients may enable the pharmaceutical compositions to be formulated into tablets, pills, capsules, liquids, gels, or syrups to aid intake by the subject. A thorough discussion of pharmaceutically acceptable carriers is available in Remington's Pharmaceutical Sciences (38).
  • the pharmaceutical composition may include a therapeutically effective amount of one or more of the variants of a TNF family ligand, the dimers, the nucleotide sequences, the vectors or the host cells of the invention.
  • a pharmaceutically effective amount is an amount able to treat the disease. The actual amount will depend on a number of factors including the size, weight, age, gender, and health of an individual, and the rate of blood clearance, and will be decided by a clinical practitioner. Generally a pharmaceutically effective amount will be between lg/kg body weight and lmg/kg body weight or less.
  • the pharmaceutical composition may include an additional therapeutic component, in particular, a component useful for the treatment of osteoporosis, rheumatoid arthritis, Paget's disease or malignancy induced bone disease.
  • a component may include Denosumab, antiresorptive agents, including bisphosphonates, the selective estrogen-receptor modulator (SERM), raloxifene, calcitonin, denosumab, teriparatide, calcium or vitamin D.
  • the additional therapeutic component may be useful for the treatment of cancer, and may include chemotherapeutics, ERMs, SERMs, or specific biological targeted therapies e.g. Herceptin.
  • the additional therapeutic component may be useful for the reducing inflammation any may include infliximab, etanercept, immunosupresives such as metroxate, and other anti-inflammatory agents such as NSAIDs.
  • the invention also includes any medical device which may have the pharmaceutical composition of the invention inserted into it or coated onto it.
  • Such devices include, stents, pins, rods, meshes, beads, syringes, plasters, microchips, micro fluidic devices, and stitches.
  • the invention provides a variant of a TNF family ligand, a dimer, a nucleotide sequence, a vector of, a host cell or the pharmaceutical composition of the invention for use in medicine.
  • the invention includes a variant of a TNF family ligand, a dimer, a nucleotide sequence, a vector, a host cell or a pharmaceutical composition according to the invention for use in the treatment of osteoporosis, rheumatoid arthritis, Paget's disease, malignancy induced bone disease or cancer, such as breast cancer.
  • the invention includes a method of treating osteoporosis, rheumatoid arthritis, Paget 's disease, malignancy induced bone disease or cancer such as breast cancer comprising administering a pharmaceutically effective amount of a variant of a TNF family ligand, a dimer, a nucleotide sequence, a vector of, a host cell or the pharmaceutical composition of the invention to a patient in need of treatment.
  • treatment encompasses therapy, and can be prophylactic or therapeutic.
  • a pharmaceutically effective amount is an amount able to treat the disease.
  • the actual amount will depend on a number of factors including the size, weight, age, gender, health of an individual, and the rate of blood clearance, and will be decided by a clinical practitioner.
  • a pharmaceutically effective amount will be between lg/kg body weight and 1 mg/kg body weight or less.
  • the invention includes the use of a variant of a TNF family ligand, a dimer, a nucleotide sequence, a vector, a host cell or a pharmaceutical composition according to the invention in the manufacture of a medicament for the treatment of osteoporosis, rheumatoid arthritis, Paget's disease, malignancy induced bone disease or cancer such as breast cancer.
  • the variant of a TNF family ligand, dimer, nucleotide sequence, vector, host cell or pharmaceutical composition according to the invention may be used for the treatment of disease in any animal.
  • the animal may be a mammal such as a camel, dog, cat, horse, cow, pig, sheep, camelid, mouse, rat, rabbit, hamster, guinea pig, pig, sheep.
  • the mammal may be a human.
  • the variant of a TNF family ligand, dimer, nucleotide sequence, vector, host cell or pharmaceutical composition according to the invention may be administered to a patient using any one or more of a number of modes of administration which will be known to a person skilled in the art.
  • modes of administration may include parenteral injection (e.g. intravenously, subcutaneously, intraperitoneally, intramuscularly, or to the interstitial space of a tissue), or by rectal, oral, vaginal, topical, transdermal, intradermal, intrathecal, intranasal, ocular, aural, pulmonary or other mucosal administration.
  • parenteral injection e.g. intravenously, subcutaneously, intraperitoneally, intramuscularly, or to the interstitial space of a tissue
  • rectal oral, vaginal, topical, transdermal, intradermal, intrathecal, intranasal, ocular, aural, pulmonary or other mucosal administration.
  • the invention includes a method for producing a variant of a TNF family ligand comprising the steps of:
  • the most hydrophilic amino acid substitution at newly solvent exposed positions may be favoured.
  • Modifying the variant to improve its properties such as to decrease its immunogenicity or improve its pharmacokinetics.
  • modifications may include one or more of pegylation, acetylation, formylation, alkylation such as methylation, and glycosylation and other possible modifications.
  • the method may be a method of producing an inhibitory variant of a TNF family ligand.
  • Figure 1 shows a schematic view of RANK inhibitor design.
  • trimeric RANKL left
  • a dimeric variant right
  • only one intact receptor binding interface is present. While still able to bind to RANK it is unable to form the biological active RANKL-RANK signaling complex.
  • Figure 2 is a schematic view depicting the binding of a TNF family ligand trimer to 3 receptors (left), and the binding of a TNF family ligand dimer to a single receptor at the only cleft formed between two ligand monomers (right).
  • the orientation of the ligand receptor complex is similar to figure la.
  • the receptors are depicted as blue circles and the TNF ligand monomers as red, blue and green ribbons.
  • Figure 3 shows capillary electrophoresis of A) murine wild-type RANKL, B) dimer 1 (mDl), and C) dimer 2 (mD2).
  • Figure 4 shows biophysical characterization of the murine RANKL dimers.
  • A) Capillary electrophoresis under denaturating conditions. The electropherograms of murine flagRANKL WT (flag-mRANKL WT) (upper trace) and mD2 (lower trace) are shown. The boxed area encloses the main protein peak. B) Detailed view of the boxed area of the main protein peak. C) Size exclusion chromatography.
  • the upper trace shows the elution profile of flag-mRANKL WT (elution volume Ve: 10 ml) and the lower traces show the superimposed elution profiles of mDl , mD2 (Ve: 1 1 ml) and an mD2 version containing an additional flag-tag (Ve: 10.9 ml).
  • the vertical line depicts the elution volume of flag-mRANKL WT and the tick marks indicate the elution volumes of calibration standards (C: Conalbumin, 75 kDa; O: Ovalbumin, 43 kDa; CA: Carbonic Anhydrase 29 kDa; R: Ribo nuclease 13.7 kDa and A: Aprotinin, 6.5 kDa) and of the void volume (V 0 8 ml) of the column.
  • WT is fiag-mRANKL WT; Dl is mDl and D2 is mD2.
  • Figure 5 shows circular dichroism (CD) far UV wavelength spectrum and thermal denaturation.
  • Upper panel CD far UV wavelength spectrum (320-200 nm) of flag- mRANKL WT (WT), mD 1 (D 1) and mD2 (D2).
  • Lower panel Thermal denaturation as monitored by CD of flag-mRANKL WT, mDl and mD2. Samples were heated from 25- 90°C with a rate of 40°C/hr, unfolding was measured at 222 nm.
  • the CD signal is expressed in mean residue molar ellipticity units.
  • Figure 6 shows a surface Plasmon resonance binding curve of A) murine RANKL WT and B) RANKL mDl and C) RANKL mD2 binding to immobilized hRANK-FC receptor
  • Figure 7 shows the results of a competitive ELISA using RANKL dimer mD2.
  • Figure 8 shows osteoclasto genesis determination by TRAP (tartrate resistant acid posphatase) assay using A) lOOng/ml wild-type mRANKL, B) lOOng/ml RANKL dimer 2 (mD2), C) lOOng/ml wild-type mRANKL and lOOng/ml RANKL dimer 2 (mD2), and D) untreated.
  • TRAP heartrate resistant acid posphatase
  • Figure 9 is a graphical representation of the results of Figure 8, A) mDl +/- mRANKL WT and B) mD2 +/- mRANKL WT.
  • Figure 10 shows A) Molecular weight determination of flag-mRANKL WT, mDl and mD2 by size exclusion chromatography using known calibration standards. Plotted is Kav of sample or standard versus the logarithm of the molecular weight (Mr), with Ka V defined as the ratio between (elution volume of protein - column void volume) and (column volume - column void volume). The Mr of flag-mRANKL WT, mDl and mD2 were obtained by interpolation of the individual Ka V 's and the calibration line.
  • Figure 12 shows a circular dichroism (CD) temperature scan of A) mDl , and B) mD2. Additional CD temperature scan data with WT control and additional replicates of mDl and mD2 can be found in the lower panel of figure 5.
  • CD circular dichroism
  • Figure 13 shows analytical gel filtration chromatography for A) murine flag wild-type RANKL, B) RANKL mD2, and C) mD2 containing an additional N-terminal flagtag. Additional gel filtration chromatography data containing mDl can be found in figure 4 C and figure 10 A.
  • Figure 14 shows analytical gel filtration chromatography, where F is flag wild-type RANKL, E and C are RANKL mD2 and D is mD2 containing an additional N-terminal flagtag.
  • Figure 14 is based on figure 13 but having all elution profiles overlaid in a single figure. Additional gel filtration chromatography data containing mDl can be found in figure 4 C and figure 10 A.
  • Figure 15 shows kinetics of RANK binding as measured by SPR.
  • the SPR sensorgrams show binding of murine RANKL WT, mDl or mD2 to mRANK-Fc at a low capture density (A) or a high capture density (B).
  • Sensorgrams in A represent measurements that were performed using a very low density of mRANK-Fc (7.5, 45 and 38 RU for WT, mDl and mD2, respectively) to ensure a 1 : 1 interaction between ligand and receptor (see text).
  • Measurements in B were performed at a high mRANK-Fc density (1880, 1942 and 1935 RU for WT, mDl and mD2 respectively).
  • Figure 16 Binding of murine RANKL dimers towards human RANK. Binding of 0.3 to 15 nM of murine RANKL WT, mDl or mD2 towards A) mRANK-Fc or B) hRANK-Fc as measured by SPR using a single cycle kinetics method. Ligand was injected with 5 consecutive injections from the lowest to the highest concentration and after the last injection dissociation was monitored for 600 s. Binding data was fitted using a 1 : 1 interaction model. WT is flag-mRANKL WT, Dl is mDl and D2 is mD2.
  • Figure 17 shows ELISA of murine RANKL WT, mDl and mD2 binding to immobilized
  • Figure 19 shows A) RANKL WT-mediated osteoclasto genesis. Formation of multinucleated cells as observed by TRAP staining in murine RAW 264.7 cells upon treatment with 100 ng/mL mRANKL WT (WT); untreated cells (NT). Multinucleated cells are marked with arrows. B & C) Inhibition of RANKL-mediated osteoclasto genesis by mDl and mD2. The relative number of multinucleated RAW 264.7 cells upon treatment with mRANKL WT alone or in the presence of 50 ng/mL and 100 ng/mL mDl
  • Figure 20 shows the conservation between human and murine RANKL. 143 of the 149 residues (89%) are identical.
  • Figure 21 A shows the sequence conservation between the human and murine RANKL sequences of dimer 1 (mDl). Mutations in dimer A are shown in dark blue, mutations in dimer B are shown in light cyan, and mutations shared between dimers A and B are shown in dark green.
  • Figure 2 IB shows the sequence conservation between the human and murine RANKL sequences of dimer 2 (mD2)_. Mutations in dimer A are shown in dark blue, mutations in dimer B are shown in light cyan, and mutations shared between dimers A and B are shown in dark green.
  • Figure 22 shows sequence alignment of the extracellular domain of murine RANKL and human RANKL. conserveed positions are depicted as dots, amino acids that differ between the murine and human the sequence are indicated in red. Positions that are mutated in mDl (A) and mD2 (B) are boxed and the substituted residues are shown above the alignment. This figure is a more concise figure based on figures 21a and b.
  • Figure 23 shows the non-conserved residue positions (red) between the human and murine RANKL sequences mapped on the dimer structure.
  • A) shows the conserved trimer interface between the human and murine RANKL sequences as mapped on the dimer structure and B) shows the conserved receptor binding interface between the human and murine RANKL sequences as mapped on the dimer structure.
  • the position of the receptor binding interface between the two adjacent monomer is indicated by the blue arrow.
  • Figure 24 shows conformation of stoichiometry by Native Gel for Dimer 1 (mDl) and 2 (mD2). Making use of a NativePAGETM Novex® 4-16% Bis-Tris Gel samples were loaded and the size was checked. Visible is one band at the expected size and two bands higher, the sizes do not correspond to the size of a trimer or monomer. A more complete version from a subsequent experiment containing WT control can be found in figure IOC.
  • Figure 25 shows conformation of stoichiometry by DLS for mD l and mD2.
  • the first peak has a polydispersity of 35% for a measured radius of 2.8 nm corresponding to a 38 kDa protein, the second and third peak correspond to a radius which is to big to be a protein.
  • the first peak has a polydispersity of ⁇ 0.1%> for a measured radius of 3.0 nm corresponding to a 46 kDa protein
  • the second and third peak correspond to a radius which is too large to be a protein.
  • Figure 26 shows representative curves of receptor binding to hRANK-Fc of wt- mRANK-L trimer and RANK-L Dimer 1 (mDl) and Dimer 2 (mD2) as determined with SPR. Concentration dependent association was reached for both dimers and for wildtype. The maximum response units are as expected for the interaction between dimeric RANK-L and one monomeric unit of mRANK-FC, as can be calculated with the formula ⁇ [MWbound/MWcaptured] x RU RANK-FCcaptured ⁇ which gives the expected maximum RU value. In the experiment with RANK-L Dimer 1 (mDl) 57 RU of mRANK-FC was captured, the expected maximum RU value would be 34.
  • Figure 27 shows binding of mRANKL WT, mDl and mD2 to hOPG as shown by SPR.
  • Directly immobilized OPG on a CM5 chip concentration range RANKL (4 nM - 31.25 pM). Concentration dependent binding, almost no dissociation.
  • the curves show the association phase, followed by the dissociation phase, regeneration of the bound ligand from the receptor and return to baseline.
  • Figure 28 shows the results of an ELISA to measure the binding of mRANKL- WT , Dl (mDl) and D2 (mD2) to A) human RANK-Fc and B) human OPG-Fc.
  • Figure 29 shows dimer l(mDl) treated cells (concentration 100 ng/mL), showing the formation of bigger osteoclasts.
  • Figure 30 shows the association of wild-type TNF ligands with their cognate receptor, when ligands bind either A) in the cleft formed between two receptors, or B) to the surface exposed surface of the receptor, wherein the large blue ovals represent receptor monomers and the small red ovals represent ligand monomers.
  • Figure 31 shows an alignment of the extracellular ligand binding domain of various TNF ligands against murine RANKL. Structurally equivalent positions of mRANKL mutations are shown in bold and underlined. All sequences shown are based on the X- ray structures, except human APRIL which is based on a homology model with murine April as template.
  • Figure 32 shows a structural alignment of the extracellular ligand binding domain of various TNF ligands against murine RANKL. All sequences shown are based on the X- ray structures, except human APRIL which is based on a homology model with murine April as template. mRANKL is in black and the amino acid positions used to create the mRANKL dimers are indicated. In grey are the other TNF-ligands.
  • Figure 33 Soluble expression of human dimeric RANKL variants in E. coli BLR (DE3) upon induction with IPTG.
  • Cells were grown in LB at 37° C while shaking at 220 rpm, upon reaching an OD600 protein expression was induced with 1 mM IPTG and temperature was lowered to 30° C.
  • Samples were recovered 4 and 16 hrs (overnight) post induction. Proteins were extracted using Novagen Bugbuster according to the suppliers specifications and insoluble and soluble fraction were separated by centrifugation for 30 min at 20.000 g. Proteins in the soluble fraction were separated on a 4-12% NuPAGE gel (Invitrogen) and transferred by Western blot.
  • histag containing proteins were detected using an anti-histag antibody (Sigma). Soluble expression could be demonstrated for all variants. From each variant 3 independent clones were tested. Of note, the signal from hRANKL D2.3 gave such a strong signal that it became overexposed.
  • Figure 34 A Purity of purified human RANKL dimer variants hD l-3 and hD2-3 as determined by SDS-PAGE/CBB stain. Proteins were purified using an Histrap column purification (HIS-TRAP: GE Lifesciences) step followed by a Gelfiltration step (Hiload 16/60 Superdex 75, GE Lifesciences). B) Difference in molecular weight as determined by a difference in elution volume between trimeric murine flagtag RANKL WT and human dimeric histag RANKL variant hD2-3 as measured on a Hiload 16/60 superdex 75 column (GE Lifesciences). Variant hD2-3 elutes at a later volume than trimeric flagtag RANKL WT and similar to murine dimeric RANKL mDl and mD2.
  • FIG. 35 Binding of dimeric human RANKL variants and human RANKL WT towards murine RANK at 37° C (left) or human RANK (right) receptor.
  • Receptors were captured at a level of 200 RU using a Protein A (Sigma) modified surface of a CM5 chip (Biacore GE Lifesciences).
  • Receptor binding curves of purified human hDl .3 and hD2.3 variants and human RANKL WT (E. coli produced, R&D systems) were recorded in real time using the single cycle kinetics method (Biacore) using a Biacore T100 (Biacore, GE Lifescience).
  • Ligands were injected in 5 consecutive injection from 1 nM to 100 nM. After subtracting the signal from the reference channel and buffer injections the resulting binding curves were fitted to a 1 : 1 interaction model using the BiaEvaluation software (Biacore GE Lifescience). Kinetic constants and affinity constants are shown in table 11. Flow rate was 50 ul/min and protein A surface was regenerated using 10 mM Glycine pH 2 solution, murine RANK-Fc and human RANK-Fc were obtained from R&D systems. Running buffer was HBS-EP+ (Biacore GE Lifescience).
  • RANKL like most other TNF ligand family members, binds the RANK receptor in the cleft between two adjacent monomers and RANKL induced trimerization of the RANK receptor results in activation of downstream signalling.
  • a competitive antagonist could be created that binds RANK receptor but is unable to trimerize and activate it.
  • mDl mouse Dimer 1
  • mD2 mouse Dimer 2
  • dimer 1 was predicted to destabilize trimer formation by ⁇ 8 kcal/mol. Trimer destabilization by dimer 2 was predicted to be ⁇ 5 kcal/mol. FoldX calculation also showed that dimer 2 was 2.8 kcal mol more stable than dimer 1 and that the propensity to form homodimers was also energetically unfavourable (table 3).
  • AAG stability energies are calculated for the heterodimers Dl (mDl) and D2 (mD2) (consisting of a monomer A and B).
  • stability energies for putative Dl and D2 homodimers are also depicted, showing that these homodimers are less energetically favorable than the heterodimers.
  • Stability energies are in kcal/mol.
  • E.coli BL21 BLR A recombinant negative E.coli BL21 derivate, E.coli BL21 BLR, was transformed with the expression vector encoding mDl and mD2.
  • the E.coli BL21 BLR cells were grown in 2xLB medium at 37°C and 250 rpm until OD 0.5 was reached. Streptomycin (100 ug/mL) (Duchefa) or spectomycin (100 ug/mL) (Duchefa) was used as selective agent. After reaching OD 0.5 the cultures were induced with 1 mM Isopropyl ⁇ -D-l- thiogalactopyranoside (IPTG) (Duchefa) and the temperature was lowered to 20°C.
  • IPTG Isopropyl ⁇ -D-l- thiogalactopyranoside
  • the cells were incubated overnight and subsequently harvested by centrifugation at 10,000g for 15 minutes. The pellet was washed with PBS and resuspended in 20 mM sodium phosphate (Merck) buffer, containing 10% glycerol (Duchefa), 150 mM NaCl (Merck), and 7 mM ⁇ -mercaptoethanol (Sigma) , pH 7.5 (buffer A).
  • 20 mM sodium phosphate (Merck) buffer containing 10% glycerol (Duchefa), 150 mM NaCl (Merck), and 7 mM ⁇ -mercaptoethanol (Sigma) , pH 7.5 (buffer A).
  • the supernatant was loaded on a Ni Sepharose HP Column (HisTRAP HP GE Healthcare). The column was washed with buffer A and the his-tag containing dimer was eluted with a gradient 0-100% of elution buffer (buffer A containing 1 M imidazol (Sigma) pH 7.5). By having only monomer A tagged and using a His-tag purification step, presence of B can only be explained by complex formation between A and B. Fractions containing his-tag protein were combined, concentrated and loaded on a gelfiltration column (Superdex 75 16/60 GE Healthcare). Buffer A was used as elution buffer. Purified proteins were >90%> pure as determined using a collodial Coomassie Brilliant Blue-stained SDS-PAGE gel. Purified protein solutions were flash frozen in liquid nitrogen and stored at -80°C.
  • Circular Dichroism and thermal decay experiments were performed as described before (26). Samples were dialyzed against 20 mM Sodium phosphate buffer containing 250 mM NaCl and 10% glycerol pH 7.3 and were measured at 0.2 mg/mL.
  • Circular dichroism measurements showed that the far UV (320-200 nm) CD spectrum of the dimers is similar to the CD spectrum of flag-mRANKL WT and indicated that both dimers are properly folded and have secondary structures consistent with an all ⁇ -sheet protein (Figure 5 top panel and figure 11). Heat induced unfolding showed that both variants were structurally stable up to 60 °C (figure 5 lower panel and figure 12). mD2 as predicted by FoldX appears to be slightly (3-5° C) more stable than mDl ( Figure 5).
  • Analytical gelfiltration was performed with 100 sample with a concentration of 0.6 mg/mL. Samples were run 10/30 75 (GE Healthcare). The column was calibrated with protein standards of known size. Buffer A was used as elution buffer with a flow rate of 0.5 or 1 mL/min. The retention time was related to the retention times of the calibration standards, as shown in Table 4, below.
  • dRANKL D2 is mD2; fiagRANKL WT is murine fiagRANKL WT
  • Samples were also analyzed by Dynamic Light Scattering (DLS). Purified samples (0.5- 2 mg/mL) were before measurements filtered over a 0.2 ⁇ filter. 20 measurements per sample were recorded. Data analysis was performed using Dynapro software, the data was analyzed as being bimodal. Blue native gel electrophoresis (NativePage 12% Invitrogen) was performed according to manufacturers instructions.
  • DLS Dynamic Light Scattering
  • the calculated values from the DLS experiment are 38 kDa and 46 kDa for dimer 1 (mDl) and dimer 2 (mD2), respectively (Figure Figure 4D and Figure 25). While RANKL dimer 2 shows a minimal polydispersity ( ⁇ 0.1%), the polydispersity of dimer 1 was considerably larger at 35%. This is in line with the possible aggregation as observed in the BN-PAGE experiment described above. As for size exclusion chromatography, the mass calculated in the DLS experiments depends on the shape of the protein and this might explain why the values are slightly higher than would be expected (29). Example 4 - binding of RANKL variants Dl and D2 to only one receptor molecule
  • a Surface Plasmon Resonance receptor binding assay was used as a shape independent method to determine the molecular weight of the RANKL dimers.
  • this assay In contrast to GFC and DLS which measure the oligomerization state of relatively concentrated samples (> 100 ug/ml), this assay has the added benefit that it measures the MW; and hence the oligomerization state, at physiologically relevant concentrations ( ⁇ 1 ug/ml).
  • the maximum response as recorded in a SPR assay depends on the MW and the amount (in response units [RU]) of the receptor coupled to the sensor chip and the MW of the analyte when observing a 1 : 1 binding (Equation 1).
  • T Re ceptor Binding parameters were determined using a Surface Plasmon Resonance based assay on a Biacore200 system (GE Healthcare).
  • a CI chip (GE Healthcare) was coated with 1000 response units of protein A (Sigma) using standard amine coupling chemistry as described before (15,27,28).
  • the calculated R max for dimeric RANKL variant mD l at a receptor capture level of hRANK-Fc of 57 RU is 34 RU and for dimeric variant mD2 at a capture level of 25 RI is 15 RU.
  • the measured levels are 30 RU and 14 RU, respectively.
  • the measured R max for mRANKL wild-type is (65 RU) at a capture level of 37 RU.
  • the measured values of the dimers and wt closely agree with the calculated theoretical values.
  • the oligomerization state of the RANKL variants is dimeric ( Figure 26).
  • Table 5 Determination of the stoichiometric ratio of the interaction between murine RANKL WT (WT), mDl or mD2 with immobilized murine RANK-Fc.
  • WT murine RANKL WT
  • mDl and mD2 form a 1 : 1 complex with RANK-Fc while at high receptor density the RANKL WT/ RANK-Fc interaction approach a stoichiometric ratio of 1 :3.
  • the stoichiometric ratio's for the interactions between mDl or mD2 and RANK remain around 1 :1.5.
  • the following molecular weights were used for the calculation: 56.3, 36.9, 37 and 60 kDa for murine WT, mDl, mD2 and mRANK-Fc respectively.
  • the dimers mDl and mD2 also were still able to bind to (A) murine RANK-Fc and (B) murine OPG-Fc (figure 17A and B).
  • hRANK-FC (Sigma) was captured in flow cell 2 at a level of 40 RU by a 60 s injection pulse of a 0.7 ug/ml hRANK-FC solution at a flow rate of 50 uL/min.
  • Flow cell 1 served as reference cell.
  • serial dilutions ranging from 562 nM to 100 pM of murine RANKL wt (R&D systems), or the purified RANKL dimers mDl and mD2 were injected over both flow cells at 50 uL/min and 37°C. Binding was measured in real time.
  • hOPG-Fc (R&D systems) was immobilized directly on the surface of a CM5 chip (GE Healthcare) using a standard amine coupling procedure according to manufacturer's instructions.
  • RANKL dimers mDl and mD2 or murine RANKL WT were injected at 4 nM at a flow of 70 uL/min and binding was measured in real time. The temperature was set at 25°C during the measurements. Between cycles the surface was regenerated with a 30s injection of 10 mM Glycine, pH 1.5. Association and dissociation was visualized with BIAevaluation software from Biacore (figure 27).
  • K D dissociation constants
  • association rate constants were ⁇ 5 fold (dimer 1) and ⁇ 10 fold (dimer 2) faster than the association rate constant of wild-type murine RANKL, as shown in Table 6, above.
  • the binding off-rate constant (k 0ff ) of wild-type murine RANKL was ⁇ 2 to 6 fold slower than for dimer 1 and dimer 2. SPR binding measurements to OPG at 25°C showed similar results as RANK binding (Figure 27B).
  • Figure 15A shows typical sensorgrams of flag-mRANKL WT binding to only 7.5 RU of captured RANK-Fc. Maximum binding of 5.5 RU is reached, which is slightly lower than the expected R max value of 6.8 RU for a 1 : 1 ratio, but definitely larger than 3.4 RU, the R max value expected for a binding ratio of 2 molecules of RANK per flag-mRANKL WT trimer. Similar to mRANKL WT, binding of RANKL mDl or mD2 to mRANK-Fc results in slightly lower R max values than expected for a 1 : 1 binding ratio: 24 RU D l compared to 26 RU, and 20 RU D2 compared to 23 RU ( Figure 15 A).
  • binding ratios could be reached with mDl and mD2 at densities of mRANK-Fc of ⁇ 50 RU.
  • the approximate 1 : 1 binding of RANKL proteins to mRANK-Fc allows fitting the data to the 1 : 1 Langmuir binding model.
  • the association rate constants (k a ) are comparable for flag-mRANKL WT, mDl and mD2 (Table 2).
  • the dissociation rate constants ( 3 ⁇ 4) differ more, but also have large standard deviations due to the slow and thus hardly measurable dissociation.
  • the calculated K D values of the WT and Dl and D2 variants are similar and in the subnanomolar range, all showing a high affinity towards the RANK receptor.
  • Table 7 Binding kinetics of mDl, mD2 and trimeric murine RANKL WT to mRANK- Fc as shown by SPR.
  • a mean values derived from three different experiments;
  • b mean values of the three kjki or k quotients of these different experiments.
  • dimer 1 and dimer 2 were investigated using a competitive ELISA.
  • Serial dilutions of wild-type mRANKL containing an N-terminal flagtag (flagRANKL) (0-53 nM) and a fixed concentration of dimer 1 (mDl) or dimer 2 (mD2) (0, 2.7, 5.4, 10.8, 21.7 or 43.4 nM) were added to RANK-Fc coated wells. After washing off unbound RANKL, mRANK bound flag-mRANKL was detected by an anti-flag antibody.
  • Murine RAW 264.7 cells were cultured as described (19). The cells were sub-cultured every third or fourth day by a 1 : 10 dilution in fresh medium. Cells were seeded at a density of 1,000 cells/well in 100 iL in a 96 well plate. The wells bordering the edge of the plate were filled with 100 water to prevent evaporation of medium of wells containing cells. At day three the medium was changed for Alpha-Mem medium (Invitrogen) and RANKL dimers mDl or mD2 or wt-mRANKL trimer (Antigenix America) were added at 50, 100 and 150 ng/mL.
  • Alpha-Mem medium Invitrogen
  • RANKL dimers mDl or mD2 or wt-mRANKL trimer Antigenix America
  • Inhibition of wtRANKL by dimer 1 or dimer 2 was measured by adding 100 ng/mL wtRANKL and 100 ng/mL of the dimeric variant. At day 5 the medium was replaced and wild-type or variant RANKL was added at the same concentrations.
  • osteoclasto genesis was determined using the tartrate resistance acid phosphatase (TRAP) assay (Sigma) according to manufacturer's instructions. The number of osteoclasts in each well was counted. The number of osteoclasts in the reference wells was set to one and the number of osteoclasts in the treated wells was calculated relative to the reference well. This allowed comparison between independent assays having different amount of cells at the start of the assay (figure 9 and 19 B and C).
  • TRIP tartrate resistance acid phosphatase
  • Figure 8 shows the cells after staining with the multinucleated cells being marked. As can be seen, most multinucleated cells are present in the wild-type mRANKL treated sample. In contrast, the dimer treated samples show considerably less multinucleated cells and they are also less clustered than wild-type mRANKL treated cells.
  • RANKL dimer 1 (mDl) ( Figure 29) appears to be less potent blocker of osteoclasto genesis than dimer 2 (mD2), the dimer 1 treated cells contain more nuclei and the osteoclasts are bigger than the ones observed in cells treated with RANKL dimer 2.
  • the number of multinucleated cells remains similar to the untreated control samples. Importantly, treatment with RANKL dimers at a concentration of 100 ng/mL does not lead to an increase in the formation of multinucleated cells ( Figure 8B and figure 19 B and C) .
  • the RANKL dimers are potent inhibitors of wt-mRANKL induced o steo clasto genesis .
  • an EC 50 for flag-mRANKL WT of 0.6 nM ( ⁇ 0.04 nM) was obtained and for mD2 an A 2 value of 4 nM ( ⁇ 0.5 nM) was calculated, i.e. the concentration of inhibitor required to shift the dose response curve by a factor of 2. This indicates that mDl and mD2 are potent inhibitors.
  • Example 9 Design and characterization of RANKL heterodimeric variants based on human RANKL.
  • Synthetic constructs optimized for expression in E. coli were ordered from DNA 2.0 (Menlo Park, CA, USA) encoding the first monomer under control of a T7 promoter and containing an N-terminal histag encoding sequence in frame with the first monomer (monomer A) followed by the second monomer (monomer B) under control of a second T7 promoter.
  • the expression cassette of the vector is similar to the pCDF-Duet-1 bicistronic co-expression vector described in example 2 but the backbone of the vector is based on the pJexpress 41 1 protein expression vector of DNA 2.0 (Menlo Park, CA, USA) which confers kanamycin resistance and uses a pUC origin of replication.
  • the protein expression host E The protein expression host E.
  • variants hDl-3 and hD2-3 were expressed at 1 litre scale in 2xYT medium at 30°C over night after induction with 1 mM IPTG.
  • hDl .3 The elution volume of hDl .3 is identical to that of hD2.3 and hence is also a dimer.
  • a surface Plasmon based receptor binding assay revealed that hDl .3 and hD2.3 bind with high affinity towards human RANK and murine RANK and rate constants and affinity constants are similar to that of human RANKL WT (E. coli produced, R&D Systems) ( Figure 35 and table 10). Taken together, this shows that the dimeric variants based on human RANKL have similar properties as the murine RANKL based dimeric variants and hence can also be used to antagonize RANKL-RANK signalling.
  • Example 10 FoldX mutagenesis at structurally equivalent positions in other TNF- ligands.
  • the impact of amino acid substitutions at these structural equivalent positions on the structural stability of the TNF-ligand trimers was assessed with the protein design algorithm FoldX.
  • the FoldX protein design algorithm has a highly accurate force-field to calculate changes in stability energy in proteins and it shows an excellent correlation with experimentally derived energetic parameters (24, 25). For that reason, it has been many times successfully used to improve stability or affinity of various proteins (15,26,27,28).
  • High resolution structures were downloaded from the protein database (PDB).
  • the following x-ray structure files were used: 1S55 (murine RANKL); ld4v (human TRAIL); lkxg (human BAFF); lxul (murine APRIL); 4tsv (human TNFa); ltnr (human TNFP); laly (human CD40L); 2hev (human OX40L) and; 2r32 (human GITRL).
  • a homology based model of human RANKL was constructed using FoldX was constructed based on the murine structure. If necessary the biopolymer was reconstructed into the trimeric form using the transformations enclosed in the PDB file. Next, the structures were cleaned and repaired using FoldX.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Toxicology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Abstract

La présente invention concerne des variants des ligands de la famille du TNF qui ont subi une mutation au niveau de l'interface de trimérisation du ligand de manière à ce qu'ils ne soient pas capables de s'assembler en trimères, et à ce que soit ils s'assemblent en dimères, soit ils restent sous la forme de monomères. Ces ligands se lient au récepteur du TNF mais ne sont pas capables de l'activer, fonctionnant effectivement comme des inhibiteurs compétitifs. L'invention concerne en outre des acides nucléiques codant pour les variants des ligands de la famille du TNF, des vecteurs et des cellules hôtes comprenant l'acide nucléique ainsi que des procédés de traitement de maladies associées à une signalisation aberrante par un récepteur du TNF.
EP11813430.3A 2010-12-24 2011-12-23 Variants de ligands de la famille du tnf Withdrawn EP2655407A2 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP11813430.3A EP2655407A2 (fr) 2010-12-24 2011-12-23 Variants de ligands de la famille du tnf

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP10252230A EP2468764A1 (fr) 2010-12-24 2010-12-24 Variantes du ligand de la famille TNF
EP11813430.3A EP2655407A2 (fr) 2010-12-24 2011-12-23 Variants de ligands de la famille du tnf
PCT/IB2011/055937 WO2012085891A2 (fr) 2010-12-24 2011-12-23 Variants de ligands de la famille du tnf

Publications (1)

Publication Number Publication Date
EP2655407A2 true EP2655407A2 (fr) 2013-10-30

Family

ID=44202115

Family Applications (2)

Application Number Title Priority Date Filing Date
EP10252230A Withdrawn EP2468764A1 (fr) 2010-12-24 2010-12-24 Variantes du ligand de la famille TNF
EP11813430.3A Withdrawn EP2655407A2 (fr) 2010-12-24 2011-12-23 Variants de ligands de la famille du tnf

Family Applications Before (1)

Application Number Title Priority Date Filing Date
EP10252230A Withdrawn EP2468764A1 (fr) 2010-12-24 2010-12-24 Variantes du ligand de la famille TNF

Country Status (3)

Country Link
US (1) US20140096274A1 (fr)
EP (2) EP2468764A1 (fr)
WO (1) WO2012085891A2 (fr)

Families Citing this family (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2953634B1 (fr) 2013-02-07 2021-05-26 The General Hospital Corporation Procédés d'expansion ou de déplétion de lymphocytes t régulateurs
US11299528B2 (en) 2014-03-11 2022-04-12 D&D Pharmatech Inc. Long acting TRAIL receptor agonists for treatment of autoimmune diseases
CN104059141B (zh) * 2014-04-14 2017-09-12 江苏省原子医学研究所 一种小鼠rankl突变体及其表达载体的构建、表达及应用
US11111284B2 (en) 2014-08-21 2021-09-07 The General Hospital Corporation Tumor necrosis factor superfamily and TNF-like ligand muteins and methods of preparing
US9623081B2 (en) 2014-10-03 2017-04-18 Ntercept, Llc Compositions and methods for inhibiting the biological activity of soluble biomolecules
AU2016263198C1 (en) 2015-05-15 2023-10-05 The General Hospital Corporation Antagonistic anti-tumor necrosis factor receptor superfamily antibodies
CA3008392C (fr) 2015-12-17 2021-11-09 The Johns Hopkins University Amelioration de la sclerose systemique a l'aide d'agonistes de recepteurs de mort cellulaire
CA3017622A1 (fr) * 2016-03-16 2017-09-21 Merrimack Pharmaceuticals, Inc. Molecule trail modifiee pour la therapie du cancer
WO2017176762A1 (fr) 2016-04-06 2017-10-12 Nanotics, Llc Particules comprenant des sous-particules ou des échafaudages d'acide nucléique
EA201892260A1 (ru) 2016-04-07 2019-03-29 Дзе Джонс Хопкинс Юниверсити Композиции и способы для лечения панкреатита и боли с применением агонистов рецептора смерти
CN117903308A (zh) 2016-05-13 2024-04-19 综合医院公司 拮抗性抗肿瘤坏死因子受体超家族抗体
US11767353B2 (en) 2020-06-05 2023-09-26 Theraly Fibrosis, Inc. Trail compositions with reduced immunogenicity

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5447851B1 (en) 1992-04-02 1999-07-06 Univ Texas System Board Of Dna encoding a chimeric polypeptide comprising the extracellular domain of tnf receptor fused to igg vectors and host cells
US7381792B2 (en) 2002-01-04 2008-06-03 Xencor, Inc. Variants of RANKL protein
CA2472049A1 (fr) * 2002-01-04 2003-07-17 Xencor Proteines negatives dominantes et procedes associes
US20060014248A1 (en) * 2003-01-06 2006-01-19 Xencor, Inc. TNF super family members with altered immunogenicity

Also Published As

Publication number Publication date
US20140096274A1 (en) 2014-04-03
WO2012085891A2 (fr) 2012-06-28
EP2468764A1 (fr) 2012-06-27
WO2012085891A3 (fr) 2012-11-29

Similar Documents

Publication Publication Date Title
US20140096274A1 (en) TNF Family Ligand Variants
US20210122805A1 (en) Fc FUSION PROTEINS COMPRISING NOVEL LINKERS OR ARRANGEMENTS
US20200347115A1 (en) Novel tnf family ligand trimer-containing antigen binding molecules
Compaan et al. The crystal structure of the costimulatory OX40-OX40L complex
AU2011240624B2 (en) TRAIL R2-specific multimeric scaffolds
US8669350B2 (en) TNF receptor Fc fusion proteins and in vivo methods of use
AU2005318086B2 (en) BCMA polypeptides and uses thereof
CA2831820A1 (fr) Polypeptides recombines membres de la famille des ligands tnf ayant un domaine de liaison a l'anticorps et leurs utilisations
Gong et al. CD154-CD40 interactions are essential for thymus-dependent antibody production in zebrafish: insights into the origin of costimulatory pathway in helper T cell-regulated adaptive immunity in early vertebrates
NZ545221A (en) Crystalline tumor necrosis factor receptor 2 polypeptides
EP2655411A1 (fr) Protéines à domaine échafaudage à base de fibronectine qui se lient à l'il-23
AU2013223801A1 (en) Tumour necrosis factor receptor fusion proteins and methods of using the same
EP1673389A2 (fr) Nouveaux variants de la proteine cd40l
AU2004297039A1 (en) Improved cytokine design
US20050181994A1 (en) Novel variants of CD40L protein
JP6320973B2 (ja) B細胞活性化因子の拮抗物質、その調製方法及び利用法
AU2011346554A1 (en) TNF family ligand variants
US9920119B2 (en) Chimeric molecule involving oligomerized FasL extracellular domain
CA3189227A1 (fr) Proteine de fusion comprenant un domaine de liaison d'antigene et un domaine de cytokine-trimere

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20130724

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

DAX Request for extension of the european patent (deleted)
17Q First examination report despatched

Effective date: 20150727

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20170120